Skip to main content
Erschienen in: Journal of Translational Medicine 1/2018

Open Access 01.12.2018 | Research

Subtype-specific associations between breast cancer risk polymorphisms and the survival of early-stage breast cancer

verfasst von: Fangmeng Fu, Wenhui Guo, Yuxiang Lin, Bangwei Zeng, Wei Qiu, Meng Huang, Chuan Wang

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2018

Abstract

Background

Limited evidence suggests that inherited predisposing risk variants might affect the disease outcome. In this study, we analyzed the effect of genome-wide association studies—identified breast cancer-risk single nucleotide polymorphisms on survival of early-stage breast cancer patients in a Chinese population.

Methods

This retrospective study investigated the relationship between 21 GWAS-identified breast cancer-risk single nucleotide polymorphisms and the outcome of 1177 early stage breast cancer patients with a long median follow-up time of 174 months. Cox proportional hazards regression models were used to estimate the hazard ratios and their 95% confidence intervals. Primary endpoints were breast cancer special survival and overall survival while secondary endpoints were invasive disease free survival and distant disease free survival.

Results

Multivariate survival analysis showed only the rs2046210 GA genotype significantly decreased the risk of recurrence and death for early stage breast cancer. After grouping breast cancer subtypes, significantly reduced survival was associated with the variant alleles of rs9485372 for luminal A and rs4415084 for triple negative breast cancer. Importantly, all three single-nucleotide polymorphisms, rs889312, rs4951011 and rs9485372 had remarkable effects on survival of luminal B EBC, either individually or synergistically. Furthermore, statistically significant multiplicative interactions were found between rs4415084 and age at diagnosis and between rs3803662 and tumor grade.

Conclusions

Our results demonstrate that breast cancer risk susceptibility loci identified by GWAS may influence the outcome of early stage breast cancer patients’ depending on intrinsic tumor subtypes in Chinese women.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12967-018-1634-0) contains supplementary material, which is available to authorized users.
Fangmeng Fu, Wenhui Guo, Yuxiang Lin and Bangwei Zeng contributed equally to this work
Abkürzungen
GWAS
genome-wide association study
SNPs
single nucleotide polymorphisms
BC
breast cancer
EBC
early-stage breast cancer
HRs
hazard ratios
CIs
confidence intervals
BCSS
breast cancer special survival
OS
overall survival
iDFS
invasive disease free survival
DDFS
distant disease free survival
HR
hormone receptor
AJCC
American Joint Commission on Cancer
TNM
tumor-node-metastasis
ER
estrogen receptor
PR
progesterone receptor
HER2
human epidermal growth factor-2
MAF
minor allele frequency
NHGRI
National Human Genome Research Institute
ZC3H11A
zinc finger CCCH domain-containing protein 11A
HMEC
human mammary epithelial cells

Background

Breast cancer (BC) is the most common diagnosed cancer and the fifth leading cause of cancer death among women in China [1]. The 5-year survival of early stage breast cancer (EBC) patients in China is about 58–78%, which is low compared to that in American and varies in different geographic areas of China [2]. Traditionally, there are some prognostic factors for EBC survival including tumor size, lymph node involvement, tumor grade, hormone receptor (HR) status. However it has been proven that inherited host characteristics, such as single nucleotide polymorphisms (SNPs), play an important role [3].
Recently, genome-wide association studies (GWAS) have been widely applied to search genetic variations and disease association. It is worth noting that some susceptibility genes or polymorphisms identified by GWAS have been proven to not only be associated with predisposition to malignant tumors, but also influence their clinical outcome [46]. Only one study and one meta-analysis examined the relationship between GWAS-identified BC risk polymorphisms and the outcome for BC, both of which focused on Caucasian populations [6, 7]. However, rs6504950 and rs3803662 had different effects on the survival of BC patients in those two studies. Differences might be due to the different sample sizes and the different enrolled BC cases. Still, those studies already demonstrated the possible associations between BC risk loci and BC survival.
Similarly, there had been some BC-risk GWAS focusing on East Asian women and that found several BC risk variants, most of which were different from those identified in other ethnic populations [8, 9]. However, the relation between these polymorphisms and survival of EBC Asian patients has never been established. In the present study, we analyzed the association between 21 GWAS-identified SNPs and the survival of patients in Southeastern China with EBC.

Methods

Study populations

This is a hospital-based study including 1177 early breast cancer cases from Fujian Medical University Union Hospital from July 2000 and October 2014. All the participants were histopathologically confirmed with invasive breast cancer and subsequently treated with curative surgical resection and systemic therapy. Clinicopathological and demographic data were collected from the hospital records and survival data were obtained from the followed-up database which was renewed annually. The patients were staged according to the 7th version of American Joint Commission on Cancer (AJCC) tumor-node-metastasis (TNM) staging system [10]. Estrogen receptor (ER)/progesterone receptor (PR) positivity was determined by IHC analysis of the number of positively stained nuclei (≥ 10%) and hormone receptor (HR) positivity was defined as being either ER+ and/or PR+. Tumors were considered human epidermal growth factor-2 (HER2) positive when cells exhibited strong membrane staining (3+). Expressions of 2+ would require further in situ hybridization testing for HER2 gene amplification while expressions of 0 or 1+ were regarded as negative. The subtypes were categorized as follows [11]: luminal A (ER+, PR+ > 20%, HER2−, Ki67 < 14% or grade I when Ki67 was unavailable), luminal B (HR+, HER2−, Ki67 > 14% or grade II/III when Ki67 was unavailable or HR+, HER2+); HER2 enriched (HR−, HER2+) and triple negative (HR− and HER2−). The study was approved by the Institutional Ethics Committee and all participants consented to genetic testing at the time of their participation and contributed data.

SNPs selection

We selected the polymorphisms associated with breast cancer susceptibility from the US National Human Genome Research Institute (NHGRI) Catalog of Published Genome-Wide Association Studies. We used the following inclusion criteria: (i) the significance level for genome-wide association was considered to be P ≤ 1 × 10−9; (ii) the minor allele frequency (MAF) was at least 10% in the HapMap CHB data of the public SNP database (http://​www.​ncbi.​nlm.​nih.​gov/​SNP); (iii) pair wise linkage disequilibrium (LD) between the eligible SNPs calculated by Haploview 4.1 software must be less than 0.8 (r2 < 0.8). At last, 21 polymorphisms were applied in this study which can be found in Additional file 1: Table S1.

DNA extraction and SNPs genotyping

Blood samples were collected in EDTA anticoagulant tubes and stored at − 80 °C until DNA extraction. Genomic DNA was extracted using the Whole-Blood DNA Extraction Kit (Bioteke, Beijing, China), according to the manufacturer’s protocol. The genotype analysis was performed by SNPscan, which is a high-throughput SNPs genotyping technology (Genesky Biotechnologies Inc., Shanghai, China). Finally, the raw data were analyzed by the GeneMapper 4.0 Software (Applied Biosystems, Foster City, CA). 5% of samples were randomly selected as blinded duplicates for quality assessment purposes and 100% concordance was obtained.

Statistical analyses

Overall survival (OS) and breast cancer specific survival (BCSS) were our primary endpoints and defined as the time from the date of cancer diagnosis to the date of mortality for all cause and breast cancer, respectively. Disease free survival (DFS) and distant disease free survival (DDFS) were our secondary endpoints and calculated separately as the time from the date of diagnosis to the date of any recurrence and distant recurrence to the last patient contact [12]. Survival data were analyzed using the Kaplan–Meier method with the log-rank test and multivariate Cox stepwise regression analysis to the end of follow-up (2016.12.31). Adjustment for age at diagnosis, tumor size, lymph node involvement, histological grade, ER status, and HER-2/neu expression were applied. The hazard ratios (HRs) and 95% confidence interval (CI) for each factor in multivariate analyses were calculated from the Cox-regression model. The Chi square-based Q test was used to examine the heterogeneity between subgroups. The possible gene-environment interactions were also evaluated by the Cox proportional hazard regression models. All tests were 2-sided, and P values of < 0.05 were considered statistically significant. SAS 9.4 (SAS Institute Inc., Cary, NC) was used for all statistical analyses.

Results

Patient characteristics and clinical features

Patients’ clinical characteristics and survival are summarized in Table 1. All the 1177 early breast cancer cohort, were female and their mean age was 47.0 ± 10.3 years old at breast cancer diagnosis. During a median follow-up time of 174 months, 446 cases experienced recurrence (142 locoregional and 410 distant) and 343 died (333 died of BC and 10 died of other disease).
Table 1
Patients’ clinicopathological characteristics and clinical outcome
Variables
Patients
N = 1177
iDFS
DDFS
BCSS
OS
Events
LogRank P
Events
LogRank P
Events
LogRank P
Events
LogRank P
Age at diagnosis
  
0.021
 
0.087
 
0.420
 
0.402
 ≤ 35
184
85
 
76
 
59
 
61
 
 > 35
993
361
 
334
 
274
 
282
 
Tumor size (cm)
  
< 0.001
 
< 0.001
 
< 0.001
 
< 0.001
 ≤ 2
403
88
 
80
 
67
 
70
 
 > 2
774
358
 
330
 
266
 
273
 
Nodal status
  
< 0.001
 
< 0.001
 
< 0.001
 
< 0.001
 Negative
510
116
 
101
 
69
 
75
 
 Positive
667
330
 
309
 
264
 
268
 
Clinical stage
  
< 0.001
 
< 0.001
 
< 0.001
 
< 0.001
 I
257
40
 
35
 
29
 
31
 
 II + III
920
406
 
375
 
304
 
312
 
Gradea
  
< 0.001
 
< 0.001
 
< 0.001
 
< 0.001
 I + II
904
310
 
286
 
228
 
236
 
 III
271
134
 
122
 
103
 
105
 
ER
  
< 0.001
 
< 0.001
 
< 0.001
 
< 0.001
 Negative
378
177
 
165
 
149
 
150
 
 Positive
799
269
 
245
 
184
 
193
 
Variables
Patients
N = 1177
iDFS
DDFS
BCSS
OS
iDFS
DDFS
BCSS
OS
Events
LogRank P
Events
LogRank P
Events
LogRank P
Events
LogRank P
PR
  
< 0.001
 
< 0.001
 
< 0.001
 
< 0.001
 Negative
367
171
 
159
 
144
 
145
 
 Positive
810
275
 
251
 
189
 
198
 
HER2
  
< 0.001
 
< 0.001
 
< 0.001
 
< 0.001
 Negative
860
292
 
268
 
214
 
222
 
 Positive
317
154
 
142
 
119
 
121
 
Subtype
  
< 0.001
 
< 0.001
 
< 0.001
 
< 0.001
 Luminal A
236
35
 
33
 
26
 
26
 
 Luminal B
574
240
 
218
 
163
 
172
 
 HER2+
160
80
 
76
 
67
 
67
 
 Triple negative
207
91
 
83
 
77
 
78
 
aVariable including missing data
No significant difference in BC-DDFS, BCSS, and OS was shown in the subgroup of age at diagnosis (P = 0.087, 0.420, and 0.402). But patients with a tumor size > 2 cm, lymph node positive, grade III, clinical stage II + III, or HER2 positive had significantly shorter survival times, whereas being ER or HR positivity remarkably improved the survival of EBC patients (log-rank P < 0.05, Table 1). Furthermore, our intrinsic molecular subtypes (luminal A, luminal B, HER2-enriched, and triple negative) were also associated with significantly different survival (log-rank P < 0.05, Table 1).

Effects of each polymorphism on survival of EBC

Among the 21 SNPs, 6 SNPs (rs13281615, rs4415084, rs4784227, rs889312, rs10474352 and rs10816625) had a log-rank P under 0.05 in some genetic models and in some outcome indicators (log-rank P < 0.05, Table 2). But after adjusting for age at breast cancer diagnosis, tumor size, lymph node involvement, grade, hormone receptor status, and HER2 status, only rs889312 and rs2046210 had significant effect on improving survival of EBC patients. In a recessive model, rs889312 was significantly associated with better iDFS and DDFS (iDFS: adjusted HR (aHR): 0.761, 95% CI 0.583–0.994, and DDFS: aHR: 0.631, 95% CI 0.470–0.848; Table 3). Similarly, in contrast to the GG + AA genotypes, the GA genotype of rs2046210 also improve the survival of EBC patients (iDFS: aHR: 0.812, 95% CI 0.673–0.980; DDFS: aHR: 0.771, 95% CI 0.635–0.938; BCSS: aHR: 0.790, 95% CI 0.636–0.981 and OS aHR: 0.786, 95% CI 0.635–0.934, Table 3).
Table 2
Genotyping results with EBC’s survival
SNPs
Cases
WH/H/VH
iDFS (LogRank P)
DDFS (LogRank P)
BCSS (logRank P)
OS (Log Rank P)
Events
WH/H/VH
DOM
REC
COD
Events
WH/H/VH
DOM
REC
COD
Events
WH/H/VH
DOM
REC
COD
Events
WH/H/VH
DOM
REC
COD
rs10069690
789/353/34
298/139/8
0.938
0.065
0.152
273/129/8
0.689
0.128
0.221
218/107/7
0.510
0.230
0.291
225/110/7
0.533
0.191
0.257
rs13281615
293/575/308
126/196/124
0.043
0.397
0.035
112/186/112
0.178
0.619
0.241
86/154/93
0.592
0.402
0.482
89/157/97
0.531
0.320
0.362
rs13387042
932/234/11
351/91/4
0.803
1.000
0.968
322/84/4
0.767
0.830
0.944
264/66/3
0.891
0.891
0.977
274/66/3
0.664
0.934
0.898
rs1562430
801/344/32
297/136/13
0.419
0.787
0.720
272/125/13
0.363
0.516
0.600
228/97/8
0.840
0.738
0.938
234/100/9
0.940
0.955
0.996
rs2046210
361/602/214
142/220/84
0.327
0.873
0.611
134/198/78
0.180
0.964
0.361
107/162/64
0.359
0.970
0.633
112/166/65
0.231
0.829
0.481
rs2180341
715/394/68
270/147/29
0.858
0.381
0.679
245/136/29
0.556
0.136
0.326
198/115/20
0.554
0.783
0.836
204/118/21
0.547
0.676
0.809
rs2981582
493/545/139
187/204/55
0.891
0.459
0.708
173/189/48
0.843
0.843
0.945
143/149/41
0.491
0.554
0.556
146/154/43
0.581
0.459
0.547
rs3112612
776/354/46
290/140/15
0.541
0.563
0.610
263/132/14
0.263
0.660
0.391
210/110/12
0.213
0.818
0.393
218/111/13
0.290
1.000
0.545
rs3803662
532/512/133
214/185/47
0.102
0.472
0.258
193/172/45
0.309
0.795
0.594
157/138/38
0.284
0.957
0.537
165/139/39
0.141
0.946
0.309
rs4415084
392/558/226
144/204/98
0.332
0.043
0.124
130/189/91
0.256
0.038
0.106
105/152/76
0.245
0.039
0.107
110/156/77
0.345
0.059
0.160
rs4784227
550/513/113
191/211/44
0.035
0.714
0.104
177/195/38
0.077
0.905
0.164
146/155/32
0.173
0.793
0.389
148/162/33
0.091
0.773
0.235
rs889312
346/631/200
130/252/64
0.770
0.059
0.111
124/235/51
0.823
0.003
0.010
98/189/46
0.840
0.070
0.142
101/196/46
0.841
0.038
0.080
rs9485372
388/588/200
136/227/82
0.122
0.177
0.200
127/208/74
0.230
0.360
0.415
104/169/59
0.334
0.529
0.592
107/173/62
0.320
0.382
0.513
rs10474352
374/572/230
158/214/74
0.052
0.029
0.041
143/199/68
0.142
0.049
0.101
115/161/57
0.285
0.156
0.301
119/165/59
0.241
0.160
0.284
rs10816625
350/595/231
145/213/88
0.047
0.825
0.127
136/196/78
0.022
0.468
0.073
114/156/63
0.017
0.559
0.056
118/160/65
0.012
0.567
0.041
rs12922061
539/529/108
199/206/41
0.590
0.905
0.865
185/188/37
0.799
0.926
0.953
156/147/30
0.613
0.943
0.877
158/154/31
0.847
0.970
0.981
rs2290203
270/587/319
96/229/121
0.464
0.891
0.760
89/211/110
0.519
0.962
0.800
67/174/92
0.218
0.689
0.468
69/179/95
0.206
0.647
0.449
rs2296067
418/567/191
160/215/71
0.869
0.814
0.968
144/200/66
0.774
0.923
0.940
116/166/51
0.649
0.751
0.798
119/172/52
0.590
0.668
0.704
rs2981578
416/548/212
150/219/77
0.465
0.619
0.556
132/208/70
0.148
0.512
0.650
105/172/56
0.158
0.488
0.166
110/176/57
0.232
0.421
0.210
rs4951011
522/528/126
204/191/51
0.350
0.421
0.340
186/178/46
0.516
0.475
0.516
150/142/41
0.597
0.163
0.233
157/145/41
0.388
0.246
0.230
rs9693444
572/486/118
215/179/52
0.762
0.154
0.357
196/164/50
0.619
0.068
0.188
156/141/36
0.379
0.483
0.616
160/144/39
0.329
0.259
0.428
WH/H/VH wide homozygous type/heterozygote/variant homozygous type, DOM dominant model, REC recessive model, COD codominant model
Table 3
Association between the SNPs’ genotype with EBC’ survival (multivariate cox proportional hazard model)
SNPs
Cases
iDFS
DDFS
BCSS
OS
Events
Adjusted HR (95% CI)a
P value
Events
Adjusted HR (95% CI)a
P value
Events
Adjusted HR (95% CI)a
P value
Events
Adjusted HR (95% CI)a
P value
All cases
 rs889312
  CC
346
130
1 (reference)
 
124
1 (reference)
 
98
1 (reference)
 
101
1 (reference)
 
  CA
631
252
1.089 (0.880–1.347)
0.433
235
1.065 (0.856–1.326)
0.569
189
1.087 (0.850–1.389)
0.507
196
1.094 (0.859–1.393)
0.465
  AA
200
64
0.804 (0.595–1.087)
0.157
51
0.658 (0.474–0.913)
0.012
46
0.814 (0.573–1.158)
0.253
46
0.782 (0.510–1.111)
0.170
  DOM
  
1.017 (0.828–1.248)
0.876
 
0.960 (0.777–1.187)
0.706
 
1.020 (0.804–1.293)
0.872
 
1.017 (0.805–1.285)
0.887
  REC
  
0.761 (0.583–0.994)
0.045
 
0.631 (0.470–0.848)
0.002
 
0.772 (0.564–1.055)
0.105
 
0.738 (0.540–1.009)
0.057
 rs2046210
  GG
361
142
1 (reference)
 
134
1 (reference)
 
107
1 (reference)
 
112
1 (reference)
 
  GA
602
220
0.796 (0.644–0.985)
0.035
198
0.761 (0.610–0.949)
0.015
162
0.775 (0.606–0.991)
0.042
166
0.762 (0.598–0.970)
0.027
  AA
214
84
0.948 (0.722–1.244)
0.700
78
0.963 (0.727–1.275)
0.792
64
0.951 (0.696–1.299)
0.752
65
0.919 (0.675–1.250)
0.589
  DOM
  
0.833 (0.682–1.018)
0.074
 
0.809 (0.658–0.996)
0.045
 
0.818 (0.649–1.031)
0.090
 
0.800 (0.638–1.005)
0.055
  REC
  
1.094 (0.861–1.391)
0.462
 
1.142 (0.890–1.464)
0.296
 
1.116 (0.847–1.469)
0.436
 
1.089 (0.829–1.430)
0.541
  OVE
  
0.812 (0.673–0.980)
0.030
 
0.771 (0.635–0.938)
0.009
 
0.790 (0.636–0.981)
0.033
 
0.786 (0.635–0.934)
0.028
Luminal A
 rs9485372
  GG
72
10
1 (reference)
 
10
1 (reference)
 
7
1 (reference)
 
7
1 (reference)
 
  GA
124
16
0.833 (0.372–1.863)
0.656
14
0.717 (0.313–1.644)
0.432
11
0.890 (0.332–2.385)
0.817
11
0.890 (0.332–2.385)
0.817
  AA
40
9
2.201 (0.883–5.486)
0.090
9
2.192 (0.880–5.459)
0.092
8
3.280 (1.152–9.378)
0.026
8
3.280 (1.152–9.378)
0.026
  DOM
  
1.087 (0.518–2.283)
0.825
 
0.995 (0.469–2.109)
0.989
 
1.328 (0.546–3.229)
0.532
 
1.328 (0.546–3.229)
0.532
  REC
  
2.465 (1.133–5.360)
0.023
 
2.671 (1.214–5.875)
0.015
 
3.522 (1.464–8.473)
0.005
 
3.522 (1.464–8.473)
0.005
Triple negative
 rs4415084
  TT
59
24
1 (reference)
 
20
1 (reference)
 
20
1 (reference)
 
20
1 (reference)
 
  CT
83
44
1.622 (0.979–2.688)
0.061
42
1.799 (1.048–3.087)
0.033
39
1.686 (0.975–2.917)
0.062
40
1.736 (1.006–2.996)
0.047
  CC
65
23
1.785 (0.996–3.201)
0.052
21
1.813 (0.971–3.385)
0.062
18
1.549 (0.809–2.969)
0.187
18
1.551 (0.810–2.972)
0.186
  DOM
  
1.674 (1.043–2.687)
0.033
 
1.804 (1.084–3.002)
0.023
 
1.640 (0.979–2.750)
0.060
 
1.674 (1.000–2.803)
0.049
  REC
  
1.345 (0.827–2.187)
0.232
 
1.274 (0.765–2.120)
0.352
 
1.139 (0.661–1.962)
0.639
 
1.119 (0.650–1.926)
0.685
Luminal B
 rs4951011
  AA
265
120
1 (reference)
 
109
1 (reference)
 
82
1 (reference)
 
88
1 (reference)
 
  GA
253
92
0.682 (0.526–0.896)
0.006
84
0.698 (0.524–0.929)
0.014
59
0.652 (0.466–0.914)
0.013
62
0.630 (0.454–0.874)
0.006
  GG
55
28
0.883 (0.579–1.346)
0.562
25
0.888 (0.568–1.386)
0.645
22
1.025 (0.631–1.664)
0.921
22
0.965 (0.597–1.560)
0.885
  DOM
  
0.719 (0.557–0.928)
0.011
 
0.734 (0.561–0.960)
0.024
 
0.721 (0.528–0.984)
0.039
 
0.690 (0.510–0.934)
0.016
  REC
  
1.068 (0.714–1.598)
0.749
 
1.075 (0.703–1.645)
0.738
 
1.259 (0.794–1.998)
0.328
 
1.205 (0.762–1.908)
0.425
 rs889312
  CC
162
74
1 (reference)
 
70
1 (reference)
 
51
1 (reference)
 
54
1 (reference)
 
  CA
308
135
1.304 (0.778–1.374)
0.819
126
1.048 (0.782–1.406)
0.753
94
1.113 (0.790–1.568)
0.542
100
1.108 (0.794–1.546)
0.545
  AA
104
31
0.570 (0.373–0.870)
0.009
22
0.432 (0.266–0.701)
0.001
18
0.534 (0.310–0.918)
0.023
18
0.498 (0.290–0.853)
0.011
  DOM
  
0.901 (0.684–1.187)
0.459
 
0.871 (0.654–1.160)
0.344
 
0.954 (0.682–1.333)
0.781
 
0.940 (0.679–1.301)
0.708
  REC
  
0.558 (0.381–0.817)
0.003
 
0.419 (0.269–0.653)
< 0.000
 
0.498 (0.304–0.815)
0.006
 
0.465 (0.285–0.761)
0.002
Luminal B
 rs9485372
  GG
204
72
1 (reference)
 
63
1 (reference)
 
47
1 (reference)
 
49
1 (reference)
 
  GA
275
125
1.439 (1.076–1.924)
0.014
115
1.524 (1.121–2.073)
0.007
89
1.517 (1.065–2.162)
0.021
93
1.520 (1.075–2.149)
0.018
  AA
95
43
1.622 (1.111–2.370)
0.122
38
1.665 (1.116–2.485)
0.013
27
1.463 (0.910–2.350)
0.116
30
1.596 (1.012–2.516)
0.044
  DOM
  
1.482 (1.124–1.954)
0.005
 
1.557 (1.161–2.088)
0.003
 
1.504 (1.071–2.112)
0.018
 
1.538 (1.104–2.142)
0.011
  REC
  
1.307 (0.939–1.820)
0.112
 
1.294 (0.914–1.831)
0.146
 
1.137 (0.752–1.720)
0.544
 
1.239 (0.835–1.839)
0.288
DOM dominant model, REC recessive model, OVE overdominant model
aHR hazard risk, CI confidence interval; For all patients: Adjusted for age at diagnosis, tumor size, lymph node involvement, grade, hormone receptor status and Her2 status; For subtypes: Adjusted for age at diagnosis, tumor size, lymph node involvement, grade

Prognostic implication of risk variants in molecular subtypes

For a large number of patients enrolled in this study, we analyzed the association between enrolled SNPs and survival associated with different molecular subtypes of EBC. As showed in Table 3, rs9485372 and rs4415084 were still associated with a worse outcome in luminal A and triple negative EBC patients, respectively, after adjustment (for rs9485372 under the recessive model: iDFS: aHR: 2.465, 95% CI 1.133–5.360; DDFS: aHR: 2.671, 95% CI 1.214–5.875; BCSS and OS: aHR: 3.522, 95% CI 1.464–8.473; for rs4415084 under the dominant model: iDFS: aHR: 1.674, 95% CI 1.043–2.687; DDFS: aHR: 1.804, 95% CI 1.084–3.002 and OS: aHR: 1.674, 95% CI 1.000–2.803). Furthermore, in the luminal B subtype we found that rs4951011 (under the dominant model) and rs889312 (under the recessive model) could significantly improve the iDFS, DDFS, BCSS and OS of the breast cancer, while rs9485372 (under dominant model) worsens outcome (iDFS: aHR = 0.719, 95% CI 0.557–0.928, DDFS: aHR = 0.734, 95% CI 0.561–0.960, BCSS: aHR = 0.721, 95% CI 0.528–0.984 and OS: aHR = 0.690, 95% CI 0.510–0.934 for rs4951011; iDFS: aHR = 0.558, 95% CI 0.381–0.817, DDFS: aHR = 0.419, 95% CI 0.269–0.653, BCSS: aHR = 0.498, 95% CI 0.304–0.815 and OS: aHR = 0.465, 95% CI 0.285–0.761 for rs889312 and iDFS: aHR = 1.482, 95% CI 0.124–1.954, DDFS: aHR = 1.557, 95% CI 0.161–2.088, BCSS: aHR = 1.504, 95% CI 1.071–2.112 and OS: aHR = 1.538, 95% CI 1.104–2.142 for 9485872, Table 3). However, no significant effect was observed in the HER2-enriched subtype in any model of the 21 polymorphisms.

Combined analysis of three risk SNPs on survival of luminal B EBC

To assess the combined effects on risk of recurrence and death from luminal B EBC, we combined the risk genotypes of rs4951011, rs889312 and 9485372. According to the number of combined risk genotypes, the univariate survival analysis show that all of iDFS, DDFS, BCSS and OS were significantly different among different groups with different combined risk genotypes (P Log-rank < 0.01) (Fig. 1). As shown in Table 4, compared to subjects with one or no unfavorable genotype, subjects carrying more unfavorable loci had shorter survival time and had a 1.534–1.645 fold increased risk of recurrence and/of death even after adjustment (iDFS: aHR = 1.534, 95% CI 1.288–1.827, DDFS: aHR = 1.632, 95% CI 1.356–1.964, BCSS: aHR = 1.570, 95% CI 1.267–1.944 and OS: aHR = 1.645, 95% CI 1.334–2.029, respectively for trend).
Table 4
Cumulative effect of unfavorable genotypes in luminal B subtype breast cancer
Number of risk genotypesa
Cases
iDFS
DDFS
BCSS
OS
Events
Adjusted HR (95% CI)b
P value
Events
Adjusted HR (95% CI)b
P value
Events
Adjusted HR (95% CI)b
P value
Events
Adjusted HR (95% CI)b
P value
0–1
165
49
1 (reference)
 
42
1 (reference)
 
32
1 (reference)
 
33
1 (reference)
 
2
272
123
1.912 (1.369–2.670)
1.44 × E−4
109
1.894 (1.324–2.711)
4.74 × E−4
81
1.787 (1.184–2.697)
5.70 × E−3
84
1.786 (1.192–6.678)
4.97 × E−3
3
137
68
2.431 (1.679–3.519)
2.52 × E−6
67
2.744 (1.862–4.043)
3.53 × E−7
50
2.525 (1.617–3.943)
4.61 × E−5
55
2.755 (1.786–4.251)
4.59 × E−6
Trend P
  
1.534 (1.288–1.827)
1.63 × E−6
 
1.632 (1.356–1.964)
2.18 × E−7
 
1.570 (1.267–1.944)
3.66 × E−5
 
1.645 (1.334–2.029)
3.25 × E−6
ars4951011 AA, rs889312 CC + CA and rs9485372 GA + AA were presumed as unfavorable genotypes
bHR hazard risk, CI confidence interval; Adjusted for age at diagnosis, tumor size, lymph node involvement, grade

Stratification and interaction analysis

The associations between breast cancer risk loci genotypes and EBC survival were then evaluated by stratified analysis of age at diagnosis, tumor size, lymph node involvement, grade, hormone-receptor status and HER2 status. As shown in Table 5, we found that rs4415084 and rs2981582 were associated with shorter survival of the patients who were younger (rs4415084 for age at diagnosis ≤ 35 years: iDFS: aHR = 1.792, 95% CI 1.161–2.915, DDFS: aHR = 2.172, 95% CI 1.310–3.602, BCSS: aHR = 2.250, 95% CI 1.278–3.959 and OS: aHR = 1.871, 95% CI 0.988–3.544) and with higher grade tumors (rs2981582 for grade III: iDFS: aHR = 1.666, 95% CI 1.051–2.639, DDFS: aHR = 1.682, 95% CI 1.049–2.698, BCSS: aHR = 1.783, 95% CI 1.080–2.944 and OS: aHR = 1.732, 95% CI 1.050–2.855). But rs2046210 and rs3803662 had beneficial effects on survival of the patients with larger tumor (rs2046210 for tumor size > 2 cm: iDFS: aHR = 0.757, 95% CI 0.606–0.944, DDFS: aHR = 0.732, 95% CI 0.582–0.919, BCSS: aHR = 0.713, 95% CI 0.533–0.920 and OS: aHR = 0.694, 95% CI 0.540–0.992) and with higher grade tumors (rs3803662 for grade III: iDFS: aHR = 0.588, 95% CI 0.414–0.834, DDFS: aHR = 0.586, 95% CI 0.407–0.845, BCSS: aHR = 0.479, 95% CI 0.319–0.717 and OS: aHR = 0.484, 95% CI 0.324–0.722) respectively. However, we did not find that the other SNPs affected survival in the subgroups of patients with different tumor characteristics.
Table 5
Stratification analysis of polymorphism genotypes associated with EBC survival
SNPs
Variables
iDFS
DDFS
BCSS
OS
Adjusted HR (95% CI)
P valuea
Adjusted HR (95% CI)
P valuea
Adjusted HR (95% CI)
P valuea
Adjusted HR (95% CI)
P valuea
rs4415084
Age at diagnosis
        
 ≤ 35
1.792 (1.161–2.915)
0.068
2.172 (1.310–3.602)
0.014
2.250 (1.278–3.959)
0.018
1.871 (0.988–3.544)
0.009
 > 35
1.073 (0.830–1.386)
 
1.056 (0.809–1.379)
 
1.067 (0.796–1.431)
 
0.743 (0.584–0.946)
 
rs2046210
Tumor size (cm)
        
 ≤ 2
1.277 (0.791–2.061)
0.052
1.277 (0.773–2.109)
0.048
1.558 (0.874–2.780)
0.015
1.522 (0.867–2.670)
0.012
 > 2
0.757 (0.606–0.944)
 
0.732 (0.582–0.919)
 
0.713 (0.553–0.920)
 
0.694 (0.540–0.992)
 
rs2981582
Grade
        
 I + II
0.922 (0.642–1.323)
0.048
0.791 (0.532–1.177)
0.017
0.822 (0.529–1.278)
0.023
0.872 (0.571–1.331)
0.040
 III
1.666 (1.051–2.639)
 
1.682 (1.049–2.698)
 
1.783 (1.080–2.944)
 
1.732 (1.050–2.855)
 
rs3803662
Grade
        
 I + II
1.017 (0.812–1.273)
0.010
1.096 (0.866–1.387)
0.005
1.151 (0.884–1.500)
0.000
1.075 (0.830–1.392)
0.001
 III
0.588 (0.414–0.834)
 
0.586 (0.407–0.845)
 
0.479 (0.319–0.717)
 
0.484 (0.324–0.722)
 
Adjusted for age at diagnosis, tumor size, lymph node involvement, grade, hormone receptor, HER2 status, exception for stratification factor
HR hazard risk, CI confidence interval
aHeterogeneity test for differences between groups
An interaction analysis was performed (Table 6) and statistically significant multiplicative interactions on EBC survival were found both between rs4415084 genotypes and age at diagnosis (adjusted Pint: iDFS 0.045, DDFS 0.013, BCSS 0.025 and OS 0.018) and between rs3803662 genotypes and tumor grade (adjusted Pint: iDFS 0.011, DDFS 0.001, BCSS 4.7 × 10−4 and OS 9.9 × 10−4).
Table 6
The interaction analysis between risk variants and clinicopathological parameters
SNPs
Variable
iDFS
DDFS
BCSS
OS
Adjusted HRa
P value
Adjusted HRa
P value
Adjusted HRa
P value
Adjusted HRa
P value
rs4415084
Age at diagnosis
        
 CC
 ≤ 35
1 (reference)
 
1 (reference)
 
1 (reference)
 
1 (reference)
 
 CC
 > 35
1.113 (0.739–1.676)
0.609
1.270 (0.814–1.983)
0.292
1.366 (0.829–2.249)
0.221
1.346 (0.827–2.189)
0.232
 CT
 ≤ 35
1.317 (0.797–2.176)
0.282
1.421 (0.829–2.438)
0.202
1.358 (0.733–2.516)
0.331
1.271 (0.692–2.336)
0.440
 CT
 > 35
1.090 (0.734–1.619)
0.669
1.246 (0.810–1.917)
0.316
1.373 (0.847–2.229)
0.198
1.340 (0.835–2.148)
0.225
 TT
 ≤ 35
2.013 (1.161–3.488)
0.013
2.427 (1.357–4.339)
0.003
2.505 (1.310–4.788)
0.005
2.497 (1.328–4.693)
0.004
 TT
 > 35
1.180 (0.767–1.815)
0.452
1.332 (0.836–2.124)
0.228
1.461 (0.868–2.460)
0.153
1.378 (0.826–2.298)
0.219
P for multiplicative interaction
 
0.045
 
0.013
 
0.025
 
0.018
 rs3803662
 Grade
        
  GG
  I + II
1 (reference)
 
1 (reference)
 
1 (reference)
 
1 (reference)
 
  GG
  III
1.858 (1.400–2.466)
1.8E−5
1.877 (1.394–2.527)
3.3E−5
2.134 (1.543–2.952)
4.6E−6
2.018 (1.469–2.773)
1.5E−5
  GA
  I + II
1.031 (0.814–1.306)
0.801
1.106 (0.864–1.416)
0.425
1.139 (0.862–1.505)
0.361
1.054 (0.801–1.385)
0.709
  GA
  III
1.043 (0.746–1.459)
0.804
1.014 (0.711–1.446)
0.939
0.979 (0.655–1.462)
0.917
0.946 (0.639–1.403)
0.784
  AA
  I + II
0.994 (0.684–1.443)
0.973
1.081 (0.735–1.592)
0.691
1.246 (0.820–1.893)
0.303
1.195 (0.793–1.800)
0.394
  AA
  III
1.085 (0.582–2.023)
0.798
1.245 (0.665–2.331)
0.493
1.043 (0.501–2.169)
0.911
0.983 (0.474–2.041)
0.964
P for multiplicative interaction
 
0.011
 
0.001
 
4.7E−4
 
9.9E−4
aHR hazard risk, CI confidence interval; adjusted for age at diagnosis, tumor size, Lymph node involvement, grade, hormone receptor status and HER2 status, except for the interaction factor

Discussion

In this study, we evaluated the possible relation between 21 GWAS-identified BC susceptibility germline variations and EBC clinical outcome in a large Chinese cohort of 1177 EBC cases. To the best of our knowledge, this is the first study that reports the association between GWAS-identified BC susceptibility loci and clinical outcomes in a Chinese population and it produced different results from two other American studies findings [6, 7]. The most significant and novel result of this study is that the influence of BC risk polymorphisms on the outcome of EBC depends on different intrinsic molecular subtypes, especially for luminal B breast cancer.
More recently, Zhang and his colleagues demonstrated some GWAS-identified SNPs are associated with molecular subtypes of EBC in Chinese women [13]. It has been accepted worldwide that breast cancer is a complex disease and consists of several intrinsic subtypes, which have different etiologies and prognosis [14]. By altering the related genes’ expression and/or function in key signaling pathways, we gradually realize putative SNPs may take effect on the basis of molecular subtypes, whether in risk or in clinical outcome of EBC [1517].
Loci rs889312, rs4951011, and rs9485372 play significant and independent roles in survival of luminal B breast cancer patients both individually or jointly by all of the four outcome indicators (iDFS, DDFS, BCSS and OS). Recently, MAP3K1 rs889312 has been identified as a low-penetrant risk factor for breast cancer, both for ER+ or ER− breast cancer [18]. It was also demonstrated to be an independent risk factor for poor survival in diffuse-type gastric cancer in an overdominant model [19]. However, two similar investigations failed to prove this variant was associated with BC clinical outcome [6, 7], although neither of them carried out survival analysis on the basis of BC intrinsic subtypes. From most recent available data, rs889312 (C/C) was found to be significantly associated with poor DFS, DDFS and OS among HR positive breast cancer patients [20], which was similar to our results. The MAP3K1 gene is the most important member in the MAPK signal pathway which activates the transcription of essential cancer genes [21]. But the exact mechanism as to how rs889312 can change MAP3K1 protein structure and/or function is still beyond our knowledge.
The rs4951011 located in intron 2 of the zinc finger CCCH domain-containing protein 11A (ZC3H11A) and 5′-UTR of ZBED6 gene, has been first identified as a BC susceptibility loci in East Asian [8]. In another study, it was only associated with triple negative breast cancer but not other BC subtypes [22]. For rs4951011 in the dominant model, we found that the GA + GG genotype was significantly associated with a better DFS, DDFS, BCSS and OS (aHR = 0.690–0.734). However, there was no evidence indicating a relation between this variant and clinical outcome of other malignant tumors. The data of ENCODE from human mammary epithelial cells (HMEC) suggests that rs4951011 may be located in a strong enhancer region marked by peaks of several active histone acetylation modifications (H3K4me1, H3K4me3, H3K9ac, and H3K27ac) [23]. Furthermore, it was found in colorectal cancer cell lines that repressing transcription of ZBED6 modulates expression of 10 genes, including PTBN1, WWC1, WWTR1, etc., linked to important signal pathway and tumor development depended on the genetic background of tumor cells and the transcription state of its target genes [24]. So rs4951011 may regulate expression of some important metastasis-related genes and then influence the course of breast cancer.
The SNP rs9485372 was also found to play a significant role in the clinical outcome of luminal A and luminal B breast cancer patients. For luminal A BC, rs9485372 in the recessive model had a worse iDFS, DDFS, BCSS, and OS (aHR 2.465–3.522). For luminal B BC, the GA + AA genotypes had a worse iDFS, DDFS, BCSS and OS (aHR = 1.482–1.557), compared to the GG genotype. This variant is located in Table 2  (TGF-β activated kinase 1/MAP3K7 binding protein 2) which plays a pivotal role in the TGF-β pathway and contributes to development of cancer [25]. Table 2 is near the ESR1 gene and it was found to be co-expressed with ESR1 in hepatocellular carcinoma [26]. Table 2 was found to be a mediator of resistance to endocrine therapy which is a poor prognostic indicator for HR+ breast cancer patients and is a potential new target to reverse pharmacological resistance and potentiate anti-estrogen action [27]. Therefore it is possible that the association both rs9485372 and survival of luminal A and B BC patients may be mediated by regulating estrogen signaling and the TGF-β pathway.
Two GWAS-identified BC risk loci, rs1219648 and rs13387042, were found to take effect on overall survival of EBC in Tunisians [28]. On the contrary, we failed to confirm this result in our Chinese population. We attribute this difference to the following reasons. Firstly, these two studies focused on different ethnic groups with different genetics background. Secondly, we used a much bigger sample size and longer follow-up than the other study which made our result more reliable. Finally, both of these two studies are retrospective. We used the multivariate Cox proportional hazard model to evaluate the independent effect of every SNP on survival of EBC patients while the other study just used Kaplan–Meier Curve and Log-Rank Test.
Some potential limitations of our study should be taken into consideration. First, as all patients were of Chinese origin, it is unclear whether our findings are Chinese Han population—specific or common in other populations. Second, the biological mechanism of the significant SNPs in breast cancer is still unclear. Therefore, more studies with diverse ethnic backgrounds and determination of the functional characterizations of the SNPs are warranted. Nevertheless, this is the first study with integrated clinicopathological data and long enough follow-up data to investigate the association between genetic breast cancer risk polymorphisms and survival of Asian breast cancer patients depended on intrinsic molecular subtypes.

Conclusions

Our findings indicated that breast cancer risk variants are not in general strongly associated with clinical outcome. However, we illustrated that, on the basis of molecular subtypes, there are some potential BC risk polymorphisms, which are probably novel predictors for EBC outcome in Chinese patients. Large better-designed investigations with a variety of populations, as well as functional assessments are needed to verify and extend our findings.

Authors’ contributions

FMF and CW designed the study. WHG, YXL, and WQ helped in sample collection. WQ and WHG assessed the molecular genotyping and generated the data. BWZ and MH analyzed the data. FMF wrote the manuscript. All authors read and approved the final manuscript.

Acknowledgements

We would like to acknowledge all the cases joining the study and assistance of all of the research nurses of Department of General Surgery, Fujian Medical University Union Hospital.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
Not applicable.
All participants signed an informed consent form. The Ethics Committee of Fujian Medical University Union Hospital (China) approved the study. We followed the ethical guidelines of the Declaration of Helsinki.

Funding

This work was supported by the National Nature Science Foundation (Grant Number 81302320), Fujian Provincial Natural Science Foundation (Grant Number 2015J01473) and Medical Elite Cultivation Program of Fujian, China (Grant Number 2015-ZQN-ZD-14).

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66:115–32.CrossRefPubMed Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66:115–32.CrossRefPubMed
2.
Zurück zum Zitat Fan L, Strasser-Weippl K, Li JJ, St Louis J, Finkelstein DM, Yu KD, Chen WQ, Shao ZM, Goss PE. Breast cancer in China. Lancet Oncol. 2014;15:e279–89.CrossRefPubMed Fan L, Strasser-Weippl K, Li JJ, St Louis J, Finkelstein DM, Yu KD, Chen WQ, Shao ZM, Goss PE. Breast cancer in China. Lancet Oncol. 2014;15:e279–89.CrossRefPubMed
3.
4.
Zurück zum Zitat Świerniak M, Wójcicka A, Czetwertyńska M, Długosińska J, Stachlewska E, Gierlikowski W, Kot A, Górnicka B, Koperski Ł, Bogdańska M, Wiechno W, Jażdżewski K. Association between GWAS-derived rs966423 genetic variant and overall mortality in patients with differentiated thyroid cancer. Clin Cancer Res. 2016;22:1111–9.CrossRefPubMed Świerniak M, Wójcicka A, Czetwertyńska M, Długosińska J, Stachlewska E, Gierlikowski W, Kot A, Górnicka B, Koperski Ł, Bogdańska M, Wiechno W, Jażdżewski K. Association between GWAS-derived rs966423 genetic variant and overall mortality in patients with differentiated thyroid cancer. Clin Cancer Res. 2016;22:1111–9.CrossRefPubMed
5.
Zurück zum Zitat Kang BW, Jeon HS, Chae YS, Lee SJ, Park JY, Choi JE, Park JS, Choi GS, Kim JG. Association between GWAS-identified genetic variations and disease prognosis for patients with colorectal cancer. PLoS ONE. 2015;10:e0119649.CrossRefPubMedPubMedCentral Kang BW, Jeon HS, Chae YS, Lee SJ, Park JY, Choi JE, Park JS, Choi GS, Kim JG. Association between GWAS-identified genetic variations and disease prognosis for patients with colorectal cancer. PLoS ONE. 2015;10:e0119649.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Barrdahl M, Canzian F, Lindström S, Shui I, Black A, Hoover RN, Ziegler RG, Buring JE, Chanock SJ, Diver WR, Gapstur SM, Gaudet MM, Giles GG, Haiman C, Henderson BE, Hankinson S, Hunter DJ, Joshi AD, Kraft P, Lee IM, Le Marchand L, Milne RL, Southey MC, Willett W, Gunter M, Panico S, Sund M, Weiderpass E, Sánchez MJ, Overvad K, Dossus L, Peeters PH, Khaw KT, Trichopoulos D, Kaaks R, Campa D. Association of breast cancer risk loci with breast cancer survival. Int J Cancer. 2015;137:2837–45.CrossRefPubMedPubMedCentral Barrdahl M, Canzian F, Lindström S, Shui I, Black A, Hoover RN, Ziegler RG, Buring JE, Chanock SJ, Diver WR, Gapstur SM, Gaudet MM, Giles GG, Haiman C, Henderson BE, Hankinson S, Hunter DJ, Joshi AD, Kraft P, Lee IM, Le Marchand L, Milne RL, Southey MC, Willett W, Gunter M, Panico S, Sund M, Weiderpass E, Sánchez MJ, Overvad K, Dossus L, Peeters PH, Khaw KT, Trichopoulos D, Kaaks R, Campa D. Association of breast cancer risk loci with breast cancer survival. Int J Cancer. 2015;137:2837–45.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Bayraktar S, Thompson PA, Yoo SY, Do KA, Sahin AA, Arun BK, Bondy ML, Brewster AM. The relationship between eight GWAS-identified single-nucleotide polymorphisms and primary breast cancer outcomes. Oncologist. 2013;18:493–500.CrossRefPubMedPubMedCentral Bayraktar S, Thompson PA, Yoo SY, Do KA, Sahin AA, Arun BK, Bondy ML, Brewster AM. The relationship between eight GWAS-identified single-nucleotide polymorphisms and primary breast cancer outcomes. Oncologist. 2013;18:493–500.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Cai Q, Zhang B, Sung H, et al. Genome-wide association analysis in East Asians identifies breast cancer susceptibility loci at 1q32.1, 5q14.3 and 15q26.1. Nat Genet. 2014;46:886–90.CrossRefPubMedPubMedCentral Cai Q, Zhang B, Sung H, et al. Genome-wide association analysis in East Asians identifies breast cancer susceptibility loci at 1q32.1, 5q14.3 and 15q26.1. Nat Genet. 2014;46:886–90.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Low SK, Takahashi A, Ashikawa K, Inazawa J, Miki Y, Kubo M, Nakamura Y, Katagiri T. Genome-wide association study of breast cancer in the Japanese population. PLoS ONE. 2013;8:e76463.CrossRefPubMedPubMedCentral Low SK, Takahashi A, Ashikawa K, Inazawa J, Miki Y, Kubo M, Nakamura Y, Katagiri T. Genome-wide association study of breast cancer in the Japanese population. PLoS ONE. 2013;8:e76463.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Edge SB, Compton CC. The American Joint Committee on Cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol. 2010;17:1471–4.CrossRefPubMed Edge SB, Compton CC. The American Joint Committee on Cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol. 2010;17:1471–4.CrossRefPubMed
11.
Zurück zum Zitat Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thürlimann B, Senn HJ, Panel members. Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol. 2013;24:2206–23.CrossRefPubMedPubMedCentral Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thürlimann B, Senn HJ, Panel members. Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol. 2013;24:2206–23.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Gourgou-Bourgade S, Cameron D, Poortmans P, et al. Guidelines for time-to-event end point definitions in breast cancer trials: results of the DATECAN initiative (Definition for the Assessment of Time-to-event Endpoints in CANcer trials). Ann Oncol. 2015;26:873–9.CrossRefPubMed Gourgou-Bourgade S, Cameron D, Poortmans P, et al. Guidelines for time-to-event end point definitions in breast cancer trials: results of the DATECAN initiative (Definition for the Assessment of Time-to-event Endpoints in CANcer trials). Ann Oncol. 2015;26:873–9.CrossRefPubMed
13.
Zurück zum Zitat Xu Y, Chen M, Liu C, Zhang X, Li W, Cheng H, Zhu J, Zhang M, Chen Z, Zhang B. Association study confirmed three breast cancer-specific molecular subtype-associated susceptibility loci in Chinese Han Women. Oncologist. 2017;22:890–4.CrossRefPubMedPubMedCentral Xu Y, Chen M, Liu C, Zhang X, Li W, Cheng H, Zhu J, Zhang M, Chen Z, Zhang B. Association study confirmed three breast cancer-specific molecular subtype-associated susceptibility loci in Chinese Han Women. Oncologist. 2017;22:890–4.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Sapkota Y. Germline DNA variations in breast cancer predisposition and prognosis: a systematic review of the literature. Cytogenet Genome Res. 2014;144:77–91.CrossRefPubMed Sapkota Y. Germline DNA variations in breast cancer predisposition and prognosis: a systematic review of the literature. Cytogenet Genome Res. 2014;144:77–91.CrossRefPubMed
16.
Zurück zum Zitat Song N, Choi JY, Sung H, Jeon S, Chung S, Park SK, Han W, Lee JW, Kim MK, Lee JY, Yoo KY, Han BG, Ahn SH, Noh DY, Kang D. Prediction of breast cancer survival using clinical and genetic markers by tumor subtypes. PLoS ONE. 2015;10:e0122413.CrossRefPubMedPubMedCentral Song N, Choi JY, Sung H, Jeon S, Chung S, Park SK, Han W, Lee JW, Kim MK, Lee JY, Yoo KY, Han BG, Ahn SH, Noh DY, Kang D. Prediction of breast cancer survival using clinical and genetic markers by tumor subtypes. PLoS ONE. 2015;10:e0122413.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Chan CHT, Munusamy P, Loke SY, Koh GL, Wong ESY, Law HY, Yoon CS, Tan MH, Yap YS, Ang P, Lee ASG. Identification of novel breast cancer risk loci. Cancer Res. 2017;77:5428–37.CrossRefPubMed Chan CHT, Munusamy P, Loke SY, Koh GL, Wong ESY, Law HY, Yoon CS, Tan MH, Yap YS, Ang P, Lee ASG. Identification of novel breast cancer risk loci. Cancer Res. 2017;77:5428–37.CrossRefPubMed
18.
Zurück zum Zitat Zheng Q, Ye J, Wu H, Yu Q, Cao J. Association between mitogen-activated protein kinase kinase kinase 1 polymorphisms and breast cancer susceptibility: a meta-analysis of 20 case–control studies. PLoS ONE. 2014;9:e90771.CrossRefPubMedPubMedCentral Zheng Q, Ye J, Wu H, Yu Q, Cao J. Association between mitogen-activated protein kinase kinase kinase 1 polymorphisms and breast cancer susceptibility: a meta-analysis of 20 case–control studies. PLoS ONE. 2014;9:e90771.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Wei X, Zhang E, Wang C, Gu D, Shen L, Wang M, Xu Z, Gong W, Tang C, Gao J, Chen J, Zhang Z. A MAP3k1 SNP predicts survival of gastric cancer in a Chinese population. PLoS ONE. 2014;9:e96083.CrossRefPubMedPubMedCentral Wei X, Zhang E, Wang C, Gu D, Shen L, Wang M, Xu Z, Gong W, Tang C, Gao J, Chen J, Zhang Z. A MAP3k1 SNP predicts survival of gastric cancer in a Chinese population. PLoS ONE. 2014;9:e96083.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Kuo SH, Yang SY, You SL, Lien HC, Lin CH, Lin PH, Huang CS. Polymorphisms of ESR1, UGT1A1, HCN1, MAP3K1 and CYP2B6 are associated with the prognosis of hormone receptor-positive early breast cancer. Oncotarget. 2017;8:20925–38.PubMedCentralPubMed Kuo SH, Yang SY, You SL, Lien HC, Lin CH, Lin PH, Huang CS. Polymorphisms of ESR1, UGT1A1, HCN1, MAP3K1 and CYP2B6 are associated with the prognosis of hormone receptor-positive early breast cancer. Oncotarget. 2017;8:20925–38.PubMedCentralPubMed
21.
Zurück zum Zitat Witowsky JA, Johnson GL. Ubiquitylation of MEKK1 inhibits its phosphorylation of MKK1 and MKK4 and activation of the ERK1/2 and JNK pathways. J Biol Chem. 2003;278:1403–6.CrossRefPubMed Witowsky JA, Johnson GL. Ubiquitylation of MEKK1 inhibits its phosphorylation of MKK1 and MKK4 and activation of the ERK1/2 and JNK pathways. J Biol Chem. 2003;278:1403–6.CrossRefPubMed
22.
Zurück zum Zitat Chen Y, Fu F, Lin Y, Qiu L, Lu M, Zhang J, Qiu W, Yang P, Wu N, Huang M, Wang C. The precision relationships between eight GWAS-identified genetic variants and breast cancer in a Chinese population. Oncotarget. 2016;7:75457–67.PubMedCentralPubMed Chen Y, Fu F, Lin Y, Qiu L, Lu M, Zhang J, Qiu W, Yang P, Wu N, Huang M, Wang C. The precision relationships between eight GWAS-identified genetic variants and breast cancer in a Chinese population. Oncotarget. 2016;7:75457–67.PubMedCentralPubMed
23.
Zurück zum Zitat ENCODE Project Consortium, Birney E, Stamatoyannopoulos JA, Dutta A, et al. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature. 2007;447:799–816.CrossRef ENCODE Project Consortium, Birney E, Stamatoyannopoulos JA, Dutta A, et al. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature. 2007;447:799–816.CrossRef
24.
Zurück zum Zitat Ali MA, Younis S, Wallerman O, Gupta R, Andersson L, Sjöblom T. Transcriptional modulator ZBED6 affects cell cycle and growth of human colorectal cancer cells. Proc Natl Acad Sci USA. 2015;112:7743–8.CrossRef Ali MA, Younis S, Wallerman O, Gupta R, Andersson L, Sjöblom T. Transcriptional modulator ZBED6 affects cell cycle and growth of human colorectal cancer cells. Proc Natl Acad Sci USA. 2015;112:7743–8.CrossRef
25.
Zurück zum Zitat Ikushima H, Miyazono K. TGFbeta signalling: a complex web in cancer progression. Nat Rev Cancer. 2010;10:415–24.CrossRefPubMed Ikushima H, Miyazono K. TGFbeta signalling: a complex web in cancer progression. Nat Rev Cancer. 2010;10:415–24.CrossRefPubMed
26.
Zurück zum Zitat Li J, Wang Y, Zhu Y, Gong Y, Yang Y, Tian J, Zhang Y, Zou D, Peng X, Ke J, Gong J, Zhong R, Chang J. Breast cancer risk-associated variants at 6q25.1 influence risk of hepatocellular carcinoma in a Chinese population. Carcinogenesis. 2017;38:447–54.CrossRefPubMed Li J, Wang Y, Zhu Y, Gong Y, Yang Y, Tian J, Zhang Y, Zou D, Peng X, Ke J, Gong J, Zhong R, Chang J. Breast cancer risk-associated variants at 6q25.1 influence risk of hepatocellular carcinoma in a Chinese population. Carcinogenesis. 2017;38:447–54.CrossRefPubMed
27.
Zurück zum Zitat Cutrupi S, Reineri S, Panetto A, Grosso E, Caizzi L, Ricci L, Friard O, Agati S, Scatolini M, Chiorino G, Lykkesfeldt AE, De Bortoli M. Targeting of the adaptor protein Tab 2 as a novel approach to revert tamoxifen resistance in breast cancer cells. Oncogene. 2012;31:4353–61.CrossRefPubMed Cutrupi S, Reineri S, Panetto A, Grosso E, Caizzi L, Ricci L, Friard O, Agati S, Scatolini M, Chiorino G, Lykkesfeldt AE, De Bortoli M. Targeting of the adaptor protein Tab 2 as a novel approach to revert tamoxifen resistance in breast cancer cells. Oncogene. 2012;31:4353–61.CrossRefPubMed
28.
Zurück zum Zitat Shan J, Mahfoudh W, Dsouza SP, Hassen E, Bouaouina N, Abdelhak S, Benhadjayed A, Memmi H, Mathew RA, Aigha II, Gabbouj S, Remadi Y, Chouchane L. Genome-Wide Association Studies (GWAS) breast cancer susceptibility loci in Arabs: susceptibility and prognostic implications in Tunisians. Breast Cancer Res Treat. 2012;135:715–24.CrossRefPubMedPubMedCentral Shan J, Mahfoudh W, Dsouza SP, Hassen E, Bouaouina N, Abdelhak S, Benhadjayed A, Memmi H, Mathew RA, Aigha II, Gabbouj S, Remadi Y, Chouchane L. Genome-Wide Association Studies (GWAS) breast cancer susceptibility loci in Arabs: susceptibility and prognostic implications in Tunisians. Breast Cancer Res Treat. 2012;135:715–24.CrossRefPubMedPubMedCentral
Metadaten
Titel
Subtype-specific associations between breast cancer risk polymorphisms and the survival of early-stage breast cancer
verfasst von
Fangmeng Fu
Wenhui Guo
Yuxiang Lin
Bangwei Zeng
Wei Qiu
Meng Huang
Chuan Wang
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2018
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-018-1634-0

Weitere Artikel der Ausgabe 1/2018

Journal of Translational Medicine 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.